↓ Skip to main content

Human mesenchymal stromal cells inhibit tumor growth in orthotopic glioblastoma xenografts

Overview of attention for article published in Stem Cell Research & Therapy, March 2017
Altmetric Badge

Citations

dimensions_citation
61 Dimensions

Readers on

mendeley
68 Mendeley
You are seeing a free-to-access but limited selection of the activity Altmetric has collected about this research output. Click here to find out more.
Title
Human mesenchymal stromal cells inhibit tumor growth in orthotopic glioblastoma xenografts
Published in
Stem Cell Research & Therapy, March 2017
DOI 10.1186/s13287-017-0516-3
Pubmed ID
Authors

Simone Pacioni, Quintino Giorgio D’Alessandris, Stefano Giannetti, Liliana Morgante, Valentina Coccè, Arianna Bonomi, Mariachiara Buccarelli, Luisa Pascucci, Giulio Alessandri, Augusto Pessina, Lucia Ricci-Vitiani, Maria Laura Falchetti, Roberto Pallini

Abstract

Mesenchymal stem/stromal cells (MSCs) represent an attractive tool for cell-based cancer therapy mainly because of their ability to migrate to tumors and to release bioactive molecules. However, the impact of MSCs on tumor growth has not been fully established. We previously demonstrated that murine MSCs show a strong tropism towards glioblastoma (GBM) brain xenografts and that these cells are able to uptake and release the chemotherapeutic drug paclitaxel (PTX), maintaining their tropism towards the tumor. Here, we address the therapy-relevant issue of using MSCs from human donors (hMSCs) for local or systemic administration in orthotopic GBM models, including xenografts of patient-derived glioma stem cells (GSCs). U87MG or GSC1 cells expressing the green fluorescent protein (GFP) were grafted onto the striatum of immunosuppressed rats. Adipose hMSCs (Ad-hMSCs), fluorescently labeled with the mCherry protein, were inoculated adjacent to or into the tumor. In rats bearing U87MG xenografts, systemic injections of Ad-hMSCs or bone marrow (BM)-hMSCs were done via the femoral vein or carotid artery. In each experiment, either PTX-loaded or unloaded hMSCs were used. To characterize the effects of hMSCs on tumor growth, we analyzed survival, tumor volume, tumor cell proliferation, and microvascular density. Overall, the AD-hMSCs showed remarkable tropism towards the tumor. Intracerebral injection of Ad-hMSCs significantly improved the survival of rats with U87MG xenografts. This effect was associated with a reduction in tumor growth, tumor cell proliferation, and microvascular density. In GSC1 xenografts, intratumoral injection of Ad-hMSCs depleted the tumor cell population and induced migration of resident microglial cells. Overall, PTX loading did not significantly enhance the antitumor potential of hMSCs. Systemically injected Ad- and BM-hMSCs homed to tumor xenografts. The efficiency of hMSC homing ranged between 0.02 and 0.5% of the injected cells, depending both on the route of cell injection and on the source from which the hMSCs were derived. Importantly, systemically injected PTX-loaded hMSCs that homed to the xenograft induced cytotoxic damage to the surrounding tumor cells. hMSCs have a therapeutic potential in GBM brain xenografts which is also expressed against the GSC population. In this context, PTX loading of hMSCs seems to play a minor role.

Mendeley readers

Mendeley readers

The data shown below were compiled from readership statistics for 68 Mendeley readers of this research output. Click here to see the associated Mendeley record.

Geographical breakdown

Country Count As %
Unknown 68 100%

Demographic breakdown

Readers by professional status Count As %
Student > Ph. D. Student 13 19%
Student > Bachelor 12 18%
Researcher 7 10%
Student > Doctoral Student 7 10%
Student > Master 6 9%
Other 12 18%
Unknown 11 16%
Readers by discipline Count As %
Medicine and Dentistry 14 21%
Biochemistry, Genetics and Molecular Biology 12 18%
Agricultural and Biological Sciences 9 13%
Neuroscience 8 12%
Engineering 3 4%
Other 8 12%
Unknown 14 21%